Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI

Page created by Bob Montgomery
 
CONTINUE READING
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI
International Journal of
               Molecular Sciences

Review
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and
Obesity: From NAFLD to NASH to HCC
Hyunmi Kim 1,2,† , Da Som Lee 1,† , Tae Hyeon An 1,2,† , Hyun-Ju Park 1,2 , Won Kon Kim 1,2 ,
Kwang-Hee Bae 1,2, * and Kyoung-Jin Oh 1,2, *

                                          1   Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125
                                              Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; khm7607@kribb.re.kr (H.K.); dasom89@kribb.re.kr (D.S.L.);
                                              anth0291@kribb.re.kr (T.H.A.); h18509@kribb.re.kr (H.-J.P.); wkkim@kribb.re.kr (W.K.K.)
                                          2   Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and
                                              Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
                                          *   Correspondence: khbae@kribb.re.kr (K.-H.B.); kjoh80@kribb.re.kr (K.-J.O.); Tel.: +82-42-860-4268 (K.-H.B.);
                                              +82-42-879-8265 (K.-J.O.)
                                          †   These authors contributed equally to this work.

                                          Abstract: Liver disease is the spectrum of liver damage ranging from simple steatosis called as
                                          nonalcoholic fatty liver disease (NAFLD) to hepatocellular carcinoma (HCC). Clinically, NAFLD
                                          and type 2 diabetes coexist. Type 2 diabetes contributes to biological processes driving the severity
                                          of NAFLD, the primary cause for development of chronic liver diseases. In the last 20 years, the
                                          rate of non-viral NAFLD/NASH-derived HCC has been increasing rapidly. As there are currently
                                          no suitable drugs for treatment of NAFLD and NASH, a class of thiazolidinediones (TZDs) drugs
                                for the treatment of type 2 diabetes is sometimes used to improve liver failure despite the risk of
         
                                          side effects. Therefore, diagnosis, prevention, and treatment of the development and progression
Citation: Kim, H.; Lee, D.S.; An, T.H.;
                                          of NAFLD and NASH are important issues. In this review, we will discuss the pathogenesis of
Park, H.-J.; Kim, W.K.; Bae, K.-H.; Oh,
                                          NAFLD/NASH and NAFLD/NASH-derived HCC and the current promising pharmacological
K.-J. Metabolic Spectrum of Liver
                                          therapies of NAFLD/NASH. Further, we will provide insights into “adipose-derived adipokines”
Failure in Type 2 Diabetes and
Obesity: From NAFLD to NASH to
                                          and “liver-derived hepatokines” as diagnostic and therapeutic targets from NAFLD to HCC.
HCC. Int. J. Mol. Sci. 2021, 22, 4495.
https://doi.org/10.3390/ijms22094495      Keywords: non-alcholic fatty liver disease (NAFLD); non-alcoholic steatohepatitis (NASH); hepato-
                                          cellular carcinoma (HCC); type 2 diabetes; obesity; adipokines; hepatokines
Academic Editors: Montserrat Esteve
and Maria del Mar Romeno

Received: 2 April 2021                    1. Introduction
Accepted: 23 April 2021
                                               Type 2 diabetes is the main public health problem in terms of global epidemic and
Published: 26 April 2021
                                          pandemic diseases [1,2]. It is closely related with the worldwide epidemic of obesity, and
                                          approximately 75% of type 2 diabetes is related with obesity [3,4]. The relationship between
Publisher’s Note: MDPI stays neutral
                                          type 2 diabetes and obesity is further explained by the descriptive term of “Diabesity” [5].
with regard to jurisdictional claims in
                                          Actually, the modern sedentary lifestyle contributes to weight gain by promoting excessive
published maps and institutional affil-
                                          food intake and even adding physical inactivity [6,7]. For that reason, chronic metabolic
iations.
                                          diseases such as type 2 diabetes and obesity have been increasing globally.
                                               Together with obesity and type 2 diabetes, non-alcoholic fatty liver (NAFLD) is
                                          the most common liver disease, and is observed in approximately 30% of the general
                                          population [8–10]. NAFLD is characterized by hepatic triglyceride (TG) accumulation
Copyright: © 2021 by the authors.
                                          and insulin resistance [11,12]. It is the hepatic manifestation of metabolic syndrome
Licensee MDPI, Basel, Switzerland.
                                          and is a spectrum of conditions ranging from benign hepatic steatosis to non-alcoholic
This article is an open access article
                                          steatohepatitis (NASH) [13]. That is, it is broadly categorized into non-alcoholic fatty liver
distributed under the terms and
                                          (NAFL) and NASH [14]. NAFL is marked by isolated steatosis, and NASH is characterized
conditions of the Creative Commons
                                          by steatosis, lobular inflammation (inflammatory cell infiltration), and hepatocellular
Attribution (CC BY) license (https://
                                          ballooning in the presence or absence of fibrosis [15]. NASH, the more aggressive form
creativecommons.org/licenses/by/
4.0/).
                                          of NAFLD, could develop into progressive fibrosis, and is directly associated with the

Int. J. Mol. Sci. 2021, 22, 4495. https://doi.org/10.3390/ijms22094495                                            https://www.mdpi.com/journal/ijms
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI
oping hepatocellular carcinoma (HCC), which could be a major cause of morbidity
                                                      mortality induced from liver failure (Figure 1) [8]. The prevalence of NASH is app
                                                      mately ~30% for patients with NAFLD [16–19]. Approximately 20% of NASH pat
                                                      with fibrosis progress to cirrhosis [20]. Liver cirrhosis is present in only 50% of pat
                                                      with NAFLD-related HCC [21,22]. The incidence of NAFLD-related HCC without ci
Int. J. Mol. Sci. 2021, 22, 4495                                                                                                           2 of 30
                                                      sis is approximately 8% of all HCC cases [23,24]. The incidence rate for HC
                                                      NAFLD/NASH with cirrhosis ranges from 2% to 13% (Figure 1) [25,26].
                                      risk of developing Clinically,
                                                            hepatocellularNAFLD     coexists (HCC),
                                                                                carcinoma     with type   2 diabetes
                                                                                                        which    couldandbe obesity,
                                                                                                                             a majorand   it exerts
                                                                                                                                       cause    of a syne
                                                      tic effect, leading    to more  severe   liver failures  [27].  The prevalence
                                      morbidity and mortality induced from liver failure (Figure 1) [8]. The prevalence of NASH        rate  of  NAFLD is
                                      is approximatelymated
                                                          ~30%toforbe patients
                                                                      approximately      75% in [16–19].
                                                                                 with NAFLD        patientsApproximately
                                                                                                              with type 2 diabetes
                                                                                                                                 20% ofand
                                                                                                                                         NASHabout 90% i
                                      patients with fibrosis progress to cirrhosis [20]. Liver cirrhosis is present in only 50% of2 diabete
                                                      obese  population,      which  show  the   strong  relationship   of NAFLD     with type
                                                      obesity [28–31].HCC
                                      patients with NAFLD-related          NAFLD     plays
                                                                                [21,22]. Thean   important
                                                                                              incidence       role in increasesHCC
                                                                                                           of NAFLD-related        of the incidence of ty
                                                                                                                                        without
                                      cirrhosis is approximately 8% of all HCC cases [23,24]. The incidence rate for HCC in to mo
                                                      diabetes   and   its complications    [28].  Type  2  diabetes   also exacerbates   NAFLD
                                      NAFLD/NASHvere    withforms   of NASH,
                                                              cirrhosis    rangesfibrosis,
                                                                                   from 2% andtoHCC    (Figure 1)
                                                                                                  13% (Figure    1) [25,26].
                                                                                                                     [30,32,33].

                                      Figure
                            Figure 1. Type 2 1. Type 2 and
                                             diabetes  diabetes andaggravate
                                                            obesity obesity aggravate  the progression
                                                                              the progression          of NAFLD/NASH
                                                                                               of NAFLD/NASH     to HCC.toClinically,
                                                                                                                           HCC. Clinically,
                                                                                                                                      type 2 diabetes
                            coexists with
                                      typeNAFLD,  and coexists
                                           2 diabetes  it aggravates
                                                                with NAFLD
                                                                     NAFLD,toandmore  severe forms
                                                                                   it aggravates    of NASH,
                                                                                                  NAFLD       hepatocirrhosis,
                                                                                                           to more severe formsand
                                                                                                                                 ofHCC,
                                                                                                                                    NASH,leading to a
                            metabolically worse phenotype.
                                      hepatocirrhosis, and HCC, leading to a metabolically worse phenotype.

                                           Clinically, NAFLD  HCC      is one with
                                                                    coexists   of thetype
                                                                                        most   aggressive
                                                                                            2 diabetes    andgrowing
                                                                                                               obesity, cancers     [34,35].
                                                                                                                         and it exerts        Previously, hepat
                                                                                                                                          a synergistic
                                      effect, leading tovirus
                                                          more(HCV)
                                                                  severewasliverthought
                                                                                  failuresto[27].
                                                                                              be the
                                                                                                   Theleading    cause
                                                                                                         prevalence      of of
                                                                                                                      rate  HCCNAFLD[36–38],   but recent reports
                                                                                                                                          is estimated
                                                        showed
                                      to be approximately        75%up to
                                                                        in50%    of newly
                                                                            patients   withdiagnosed       HCC patients
                                                                                               type 2 diabetes              are non-viral
                                                                                                                    and about      90% in the HCC    [39,40], There
                                                                                                                                                   obese
                                      population, which NAFLD/NASH-derived               HCC has been
                                                              show the strong relationship                   highlighted.
                                                                                                         of NAFLD      withThetypeetiology
                                                                                                                                      2 diabetesof NAFLD/NASH
                                                                                                                                                     and
                                      obesity [28–31].rived
                                                          NAFLDHCCplays is very
                                                                              ancomplex
                                                                                   important androle
                                                                                                   is related  with various
                                                                                                        in increases   of the mechanisms
                                                                                                                                incidence of such type as
                                                                                                                                                        2 cellular
                                      diabetes and itsticity,   inflammation,
                                                         complications              apoptosis,
                                                                            [28]. Type            cell cycle,
                                                                                          2 diabetes           and cell death
                                                                                                        also exacerbates    NAFLD [41,42].  It is not
                                                                                                                                       to more         easy to trea
                                                                                                                                                  severe
                                      forms of NASH,    improve
                                                          fibrosis,HCC.
                                                                      and HCCTherefore,
                                                                                   (FigureNAFLD/NASH
                                                                                              1) [30,32,33]. treatment is required for prevention of
                                           HCC is one   versible    chronic
                                                           of the most        liver diseases
                                                                            aggressive     growingsuchcancers
                                                                                                          as cirrhosis  and
                                                                                                                 [34,35].     HCC. Unfortunately,
                                                                                                                           Previously,     hepatitis C there a
                                      virus (HCV) was   FDA-approved
                                                           thought to bedrugs         and treatment
                                                                              the leading      cause ofmethods      yet. but recent reports that
                                                                                                           HCC [36–38],
                                      showed up to 50% ofInnewly   this review,
                                                                          diagnosed we HCC
                                                                                         will discuss
                                                                                                patients the   pathogenesis
                                                                                                           are non-viral   HCCof[39,40],
                                                                                                                                     NAFLD/NASH
                                                                                                                                             Therefore,and NA
                                      NAFLD/NASH-derivedNASH-derived    HCCHCC       and the
                                                                              has been           current promising
                                                                                            highlighted.                 pharmacological
                                                                                                              The etiology      of NAFLD/NASH-  therapies of NA
                                      derived HCC isNASH.          Further, the
                                                          very complex        andinitiation
                                                                                    is relatedand withprogression    of NAFLD can
                                                                                                          various mechanisms         suchbe as
                                                                                                                                             affected   by organo
                                                                                                                                                 cellular
                                                        secreted from
                                      plasticity, inflammation,             metabolic
                                                                      apoptosis,    cell organs
                                                                                         cycle, andundercellmetabolic   disturbance
                                                                                                             death [41,42].    It is notsuch
                                                                                                                                          easyastotype
                                                                                                                                                    treat2 diabete
                                      and improve HCC.  obesity    [43–45]. Therefore,
                                                               Therefore,     NAFLD/NASH    we willtreatment
                                                                                                       focus on organokines
                                                                                                                   is required that     are secretedofby the ad
                                                                                                                                   for prevention
                                                        tissues
                                      irreversible chronic        anddiseases
                                                              liver     liver, which
                                                                                  such are   critical organs
                                                                                        as cirrhosis            for the
                                                                                                         and HCC.       regulation of lipid
                                                                                                                      Unfortunately,      theremetabolism.
                                                                                                                                                  are no         We
                                      FDA-approvedprovidedrugs and  newtreatment
                                                                          insights into    “adipokines”
                                                                                      methods     yet.       and “hepatokines” that can be potential diag
                                                        tic and
                                           In this review,     wetherapeutic
                                                                     will discuss targets  in NAFLD/NASH
                                                                                      the pathogenesis             and NAFLD/NASH-derived
                                                                                                              of NAFLD/NASH            and NAFLD/       HCC. The
                                      NASH-derivedthought HCC and   to be
                                                                        theable  to be biological
                                                                             current   promisingmarkers         that can predict
                                                                                                       pharmacological               NAFLD
                                                                                                                             therapies          severity from NA
                                                                                                                                          of NAFLD/
                                      NASH. Further,tothe  HCC.
                                                              initiation and progression of NAFLD can be affected by organokines
                                      secreted from metabolic organs under metabolic disturbance such as type 2 diabetes and
                                      obesity [43–45]. Therefore, we will focus on organokines that are secreted by the adipose
                                      tissues and liver, which are critical organs for the regulation of lipid metabolism. We will
                                      provide new insights into “adipokines” and “hepatokines” that can be potential diagnostic
                                      and therapeutic targets in NAFLD/NASH and NAFLD/NASH-derived HCC. They are
                                      thought to be able to be biological markers that can predict NAFLD severity from NAFLD
                                      to HCC.
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW

Int. J. Mol. Sci. 2021, 22, 4495                                                                                                                 3 of 30

                                                        2. Non-Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatit
                                       2. Non-Alcoholic (NASH)
                                                            Fatty Liver Disease (NAFLD) and Non-Alcoholic
                                       Steatohepatitis2.1.
                                                         (NASH)
                                                             Pathogenesis of NAFLD and NASH
                                       2.1. Pathogenesis2.1.1.
                                                          of NAFLD   and NASH
                                                               An Imbalance    in Fatty Acid (FA) Metabolism
                                       2.1.1. An Imbalance in Fatty Acid (FA) Metabolism
                                                              NAFLD is the most common etiology of chronic liver diseases. NAFLD results
                                             NAFLD is excessive
                                                         the most common      etiology
                                                                   triglyceride          of chronic liverindiseases.
                                                                                  (TG) accumulation           the liverNAFLD
                                                                                                                          [11,12]. results fromthe balan
                                                                                                                                   Therefore,
                                       excessive triglyceride  (TG)  accumulation    in the liver [11,12].  Therefore,    the balance  between
                                                        tween fatty acid (FA) input and FA output is critical [46,47]. That is, NAFLD dev
                                       fatty acid (FA) when
                                                        input the
                                                                andamount
                                                                     FA output    is critical [46,47].
                                                                             of “exogenous             That
                                                                                               FA uptake        is, NAFLD
                                                                                                             (dietary   intakedevelops
                                                                                                                               and adiposewhentissue lipo
                                       the amount of “exogenous       FA   uptake  (dietary   intake  and   adipose     tissue
                                                        and “endogenous FA synthesis (DNL: de novo lipogenesis)” in the liver  lipolysis)”  andis greate
                                       “endogenous FA      synthesis
                                                        “the          (DNL:
                                                              release of      de novo
                                                                         FAs (FA         lipogenesis)”
                                                                                   oxidation,   lipolysis,inand
                                                                                                              theFAliver  is greater
                                                                                                                       secretion      thanlow
                                                                                                                                 in very    “the
                                                                                                                                               density lip
                                       release of FAs (FA    oxidation,  lipolysis, and   FA  secretion
                                                        tein (VLDL)-TG)” from the liver (Figure 2).      in   very   low   density  lipoprotein
                                       (VLDL)-TG)” from the liver (Figure 2).

                                       Figure 2.
                            Figure 2. NAFLD         NAFLD development
                                                 development    is caused by isancaused  by an
                                                                                  imbalance       imbalance
                                                                                               in the          in the intrahepatocellular
                                                                                                      intrahepatocellular   fatty acid (FA) fatty   acid Hepat
                                                                                                                                              metabolism.
                                       (FA) metabolism.
                            TG accumulation     is promoted Hepatic
                                                              when theTG FAaccumulation   is promoted
                                                                              input is greater   than thewhen    the FA
                                                                                                          FA output    ininput  is greater
                                                                                                                          the liver.       than the
                                                                                                                                     The greater     FAof FA take
                                                                                                                                                   part
                            up by liveroutput
                                         is mainly   derived
                                                in the        fromgreater
                                                       liver. The    the lipolysis
                                                                            part of of
                                                                                    FAsubcutaneous     adipose
                                                                                       taken up by liver         tissue derived
                                                                                                            is mainly   TG. Another
                                                                                                                                  from major   sourceof
                                                                                                                                        the lipolysis   of FA in th
                            liver is derived  from  de  novo  lipogenesis   that converts  excess   glucose  into  FAs. On  the  other
                                       subcutaneous adipose tissue TG. Another major source of FA in the liver is derived from de novo hand,   the consumption    o
                            FA is possible  through   the signaling  pathway    involved in  lipolysis, β-oxidation,  and  TG   secretion
                                       lipogenesis that converts excess glucose into FAs. On the other hand, the consumption of FA is     (→:  signaling   pathway
                            related with TG accumulation by FA, →: signaling pathways related with the consumption of FA).
                                       possible through the signaling pathway involved in lipolysis, β-oxidation, and TG secretion (→:
                                       signaling pathways related with TG accumulation by FA, →: signaling pathways related with the
                                                                 The release of FAs from adipose tissue and the efficiency of FA uptake by liv
                                       consumption of FA).
                                                       increased in approximately 59% of patients with NAFLD [48]. Hepatic FA uptake de
                                            The releaseonofplasma
                                                            FAs from FA adipose
                                                                         concentration
                                                                                   tissue and
                                                                                           and on
                                                                                                thehepatocellular
                                                                                                     efficiency of FAcapacity
                                                                                                                         uptakeforbyFA    uptake
                                                                                                                                      liver  are that is
                                                       mined by the59%
                                       increased in approximately       number    and activity
                                                                           of patients          of specialized
                                                                                         with NAFLD               FA transporter
                                                                                                         [48]. Hepatic   FA uptake and    carrier protein
                                                                                                                                      depends
                                                       as FA  translocase   (FAT/CD36),     FA  transport   polypeptide    (FATP),
                                       on plasma FA concentration and on hepatocellular capacity for FA uptake that is determined    and   FA binding p
                                       by the number and activity of specialized FA transporter and carrier proteins suchCD36
                                                       (FABP)   [49,50]. For  example,   hepatic   expression   of fatty acid translocase     as is mar
                                       FA translocase (FAT/CD36), FA transport polypeptide (FATP), and FA binding proteinand FAB
                                                       increased    in subjects   with   NAFLD,     and   hepatic   expression   of  FABP-4
                                       (FABP) [49,50]. closely  associated
                                                       For example,          with
                                                                        hepatic     intrahepatic
                                                                                 expression       TG accumulation.
                                                                                              of fatty acid translocase CD36 is markedly
                                                            In  approximately      26%  of patients
                                       increased in subjects with NAFLD, and hepatic expression      withofNAFLD,
                                                                                                             FABP-4 theandway   to provides
                                                                                                                            FABP-5   is closelyFA pool in
                                       associated withisintrahepatic
                                                          de novo lipogenesis     (DNL) [51]. DNL is the metabolic process that synthesizes ne
                                                                       TG accumulation.
                                                       from excess
                                            In approximately    26% glucose     [52,53].
                                                                      of patients   withItNAFLD,
                                                                                           is an important
                                                                                                    the waycontributor
                                                                                                               to providestoward
                                                                                                                             FA poolhepatic
                                                                                                                                        in liverlipid accu
                                       is de novo lipogenesis (DNL) [51]. DNL is the metabolic process that synthesizes new of two
                                                       tion in the   pathogenesis    of NAFLD     [52,53]. The  effects  result from   activation
                                       FAs from excessscription
                                                         glucose factors,
                                                                   [52,53]. sterol
                                                                            It is anregulatory
                                                                                      importantelement      binding
                                                                                                  contributor         protein-1c
                                                                                                                 toward   hepatic (SREBP-1c)
                                                                                                                                   lipid accu- and car
                                                       drate  responsive    element    binding  protein   (ChREBP),
                                       mulation in the pathogenesis of NAFLD [52,53]. The effects result from activationboosted   by  insulin
                                                                                                                                         of twoand gluco
                                       transcription factors, sterol regulatory element binding protein-1c (SREBP-1c) and important
                                                       sponses   to  dietary carbohydrates     [54,55]. They   play  a synergistically   carbo-       role
                                       hydrate responsive element binding protein (ChREBP), boosted by insulin and glucose
                                       responses to dietary carbohydrates [54,55]. They play a synergistically important role in
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI
Int. J. Mol. Sci. 2021, 22, 4495                                                                                            4 of 30

                                   the coordinated regulation of hepatic DNL. In the remaining 15% of patients with NAFLD,
                                   FA pool is derived from dietary TG, which is associated with chylomicrons [48].
                                         The most acceptable theory in the pathogenesis of NAFLD is the “two-hit” hypothe-
                                   sis [56]. The first hit is “insulin resistance” caused by excessive FA flux into the liver. The
                                   second hit is “inflammation”, associated with gut-derived endotoxin, oxidative stress, and
                                   mitochondria dysfunction. It is closely related with NAFLD progression toward NASH.

                                   2.1.2. Endotoxin Behavior
                                        NALFD and other insulin resistance-related diseases are associated with activation of
                                   innate immune system, leading to chronic inflammation [57]. Recently, gut-derived endo-
                                   toxins, such as lipopolysaccharide (LPS), have been proposed to have a critical role in liver
                                   inflammation as well as progression of chronic liver diseases [58]. Under normal conditions,
                                   endotoxin can be absorbed from the intestinal lumen into the portal vein system, and the
                                   absorbed endotoxin will be rapidly removed in the hepatic reticuloendothelial system,
                                   particularly Kupffer cells [59,60]. However, obesity, type 2 diabetes, and other nutrition
                                   and environmental factors can alter intestinal permeability for bacterial overgrowth and
                                   the resulting leaky mucosal barrier allows bacterial translocation, implicating the release of
                                   gut-derived endotoxin into the systemic circulation [61,62]. The invasive pathogens and
                                   harmful byproducts influence hepatic lipid accumulation and exacerbate pro-inflammatory
                                   and fibrotic processes [60].
                                        Recently, the role of LPS from gut microbiota in the development of NAFLD and NASH
                                   has been attracting attention [63,64]. Circulating LPS levels, small intestinal permeability,
                                   and bacterial overgrowth are increased in patients with NALFD, and these factors are
                                   associated with the severity of hepatic steatosis [63,65,66]. Livers that directly receives
                                   blood from the portal vein are the main target of LPS, also known as endotoxin, and
                                   LPS-TLR4 is one of the critical pathways for NAFLD development. In mouse models, LPS
                                   infusion triggers hepatic steatosis and hepatic insulin resistance, as well as hepatic weight
                                   gain [67]. LPS exacerbates liver injury in mice fed a methionine-choline-deficient diet [68].
                                   The LPS-binding protein (LBP)-CD14 complex activates Toll-like recpeotr4 (TLR4), which
                                   is an essential inflammatory cascade in the progression of NAFLD [69,70]. Loss of LBP
                                   attenuates inflammation-mediated liver damage [71]. TLR4 can activate NF-kB and release
                                   pro-inflammatory cytokines such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-
                                   α), and IL-6 [72]. It can also recognize damage-associated molecular patterns (DAMPs)
                                   that are released from damaged cells, and mediates FA-induced inflammation [57,73].
                                   As pharmacological therapies in NAFLD and NASH targeting the microbiome, there are
                                   IMM-24e (anti-LPS antibody), solithromycin (next-generation macrolide antbiotic), and
                                   TLR4 antagonist [74].

                                   2.1.3. Oxidative Stress
                                        Chronic oxidative stress is one of the key mechanisms leading to liver injury in NAFLD.
                                   Oxidative stress is a general event that occur in NAFLD and NASH as result of excessive
                                   production of reactive oxygen species (ROS) [75,76]. ROS and lipid peroxidation can
                                   explain most of histological features of NAFLD and NASH [77,78]. In patients with hepatic
                                   steatosis, mitochondrial ROS oxidizes hepatic fat deposits, and ROS-induced expression
                                   of Fas-ligand can induce apoptosis [77,78]. Peroxidation of and intracellular membranes
                                   can directly trigger necrosis and apoptosis [77,78]. The degree of lipid peroxidation is
                                   correlated with the severity of steatosis and can explain the association between steatosis
                                   severity and the risk of necroinflammation and fibrosis in NASH [79–81]. ROS, which
                                   plays a key player in the pathogenesis of NASH, can lead to a self-perpetuating cycle
                                   of lipid peroxidation and can further generate ROS [82]. Lipid peroxidation products
                                   alter mitochondrial DNA and activate transcription factor nuclear facto-kB (NF-kB) that
                                   upregulates TNFα [83,84]. Resultingly, it further contributes to impaired mitochondrial
                                   respiration and increased ROS formation [83,84].
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW                                                                                                                  5 of 30

Int. J. Mol. Sci. 2021, 22, 4495                                                                                                                                5 of 30

                                           Increased mitochondrial β-oxidation of FFA is an important source of ROS in NAFLD
                                    and NASH [85]. Increased FFA flux in hepatic cells during early stages of NAFLD stimu-
                                          Increased mitochondrial β-oxidation of FFA is an important source of ROS in NAFLD
                                    late  mitochondrial
                                   and NASH [85]. Increased      fatty acid
                                                                         FFAoxidation
                                                                                flux in hepatic(FAO),      and
                                                                                                       cells      it reflects
                                                                                                              during              an early
                                                                                                                        early stages           effort ofstimulate
                                                                                                                                           of NAFLD           the liver
                                    compensatoryfatty
                                   mitochondrial          mechanisms       to inhibit
                                                                 acid oxidation      (FAO),liverandfat it
                                                                                                        accumulation
                                                                                                           reflects an early andeffort
                                                                                                                                    maintain
                                                                                                                                           of the lipid
                                                                                                                                                     liverhomeosta-
                                                                                                                                                             compen-
                                    sis [12].  In   NAFLD        and   NASH,      mitochondrial           FAO    is also
                                   satory mechanisms to inhibit liver fat accumulation and maintain lipid homeostasis       increased      or  at  least   preserved[12].
                                    as NAFLD
                                   In   a compensatory
                                                     and NASH,    response.    The imbalance
                                                                       mitochondrial           FAO isbetween           mitochondrial
                                                                                                           also increased         or at leastFAO      and electron
                                                                                                                                                  preserved         as a
                                    transport chain
                                   compensatory             (ETC) will
                                                         response.     Thecontribute
                                                                            imbalanceto         ROS overproduction
                                                                                              between      mitochondrial FAO     by increased
                                                                                                                                         and electronelectron      leak-
                                                                                                                                                            transport
                                    age  from     the  ETC      [12,85,86].   ROS-induced            lipid   peroxidation
                                   chain (ETC) will contribute to ROS overproduction by increased electron leakage from          leads    to  inflammation           and
                                    hepatic
                                   the   ETCfibrogenesis           through the activation
                                                [12,85,86]. ROS-induced                              of hepatic leads
                                                                                      lipid peroxidation            stellate tocells   (HSCs) [87,88].
                                                                                                                                 inflammation          and hepatic
                                           Recently,     reliable    circulating     markers       that   can  reflect
                                   fibrogenesis through the activation of hepatic stellate cells (HSCs) [87,88].          oxidative      stress   in  patients     with
                                    NAFLD       have    been     reported.    Urinary       8-iso-prostaglandin           F2α     (8-iso-PGF2α)
                                          Recently, reliable circulating markers that can reflect oxidative stress in patients with                     is known       as
                                    a reliable    indicator      of  oxidative     stress    in vivo     [89,90],   and
                                   NAFLD have been reported. Urinary 8-iso-prostaglandin F2α (8-iso-PGF2α) is known as     soluble    NOX2-derived             peptide
                                   a(sNOX2-dp)         are also
                                      reliable indicator         of an  acceptable
                                                                    oxidative     stressmarker,
                                                                                            in vivowhich[89,90],is and
                                                                                                                    associated
                                                                                                                          solublewith       ROS generation
                                                                                                                                      NOX2-derived            peptide  by
                                    activation of are
                                   (sNOX2-dp)          NOX2, also aanmember
                                                                       acceptable of the     NADPH
                                                                                        marker,       which oxidase    family [91,92].
                                                                                                               is associated        with ROS  Elevated
                                                                                                                                                    generationlevelsby of
                                    urinary 8-iso-PGF2α
                                   activation      of NOX2, aand         serumofsoluble
                                                                     member          the NADPH  NOX2-derived           peptide[91,92].
                                                                                                           oxidase family           are considered
                                                                                                                                             Elevatedas         a relia-
                                                                                                                                                             levels    of
                                    ble indicator
                                   urinary             of oxidative
                                               8-iso-PGF2α               stresssoluble
                                                                   and serum       in chronic       inflammation
                                                                                              NOX2-derived               and metabolic
                                                                                                                     peptide     are considered diseasesas a [93–95].
                                                                                                                                                               reliable
                                    They alsoofcan
                                   indicator             be usedstress
                                                     oxidative       as markers
                                                                           in chronicof oxidative
                                                                                            inflammation  stressand
                                                                                                                  for prediction         of the severity
                                                                                                                        metabolic diseases           [93–95].ofThey liver
                                    damage      in  NAFLD        [96,97].  LPS    is an   important        outer   membrane
                                   also can be used as markers of oxidative stress for prediction of the severity of liver damage    component        of  gram-neg-
                                    ative
                                   in  NAFLDbacteria    that induces
                                                    [96,97].      LPS is accelerated
                                                                          an important       inflammation
                                                                                                 outer membrane   and oxidative
                                                                                                                            component   stressof[98,99].     Elevated
                                                                                                                                                   gram-negative
                                    levels ofthat
                                   bacteria      circulating
                                                      induces NOX2         and LPS
                                                                   accelerated           in NAFLDand
                                                                                   inflammation            patients    suggest
                                                                                                               oxidative      stressthe   potential
                                                                                                                                       [98,99].          role of
                                                                                                                                                   Elevated          gut-
                                                                                                                                                                 levels
                                    derived
                                   of           LPS in
                                       circulating     NOX2 systemic    NOX2
                                                                  and LPS          activation
                                                                             in NAFLD              [100]. suggest
                                                                                                patients     Further,the sNOX2-dp
                                                                                                                              potentiallevels
                                                                                                                                            role ofare     positively
                                                                                                                                                       gut-derived
                                    related
                                   LPS        with the NOX2
                                          in systemic        histological    grading
                                                                      activation           of steatosis,
                                                                                      [100].                  inflammation,
                                                                                                Further, sNOX2-dp              levelsballooning,
                                                                                                                                         are positivelyfibrosis,     and
                                                                                                                                                               related
                                   with
                                    NAFLD  the histological
                                                 activity score    grading
                                                                      [100]. of   steatosis, inflammation,
                                                                              Gut-derived           LPS can stimulate  ballooning,
                                                                                                                                 TLR4,fibrosis,       and NAFLD
                                                                                                                                           and TLR4-mediated
                                   activity    score [100].
                                    NOXs activation           can Gut-derived
                                                                    generate ROSLPS           can stimulateinfiltration
                                                                                        by macrophage               TLR4, and         TLR4-mediated
                                                                                                                                   [101].   It can contribute   NOXs   to
                                   activation      can generate
                                    hepatic steatosis                 ROS by
                                                              and insulin         macrophage
                                                                             resistance       [101].infiltration [101]. It can contribute to hepatic
                                   steatosis    and insulin
                                           However,                resistance
                                                           the variety           [101]. changes occurred in NAFLD are insufficient to be
                                                                          of metabolic
                                          However,
                                    explained      onlythe  by variety    of metabolic
                                                                 the “two-hit”                changes
                                                                                     hypothesis.            occurred
                                                                                                         Most            in NAFLD
                                                                                                                 metabolic                are insufficient
                                                                                                                                  disorders     such as obesity,  to be
                                   explained      only bymetabolic
                                    type 2 diabetes,           the “two-hit”      hypothesis.
                                                                           syndrome,          and Most       metabolicare
                                                                                                     dyslipidemia            disorders
                                                                                                                                the risksuchfactorsas obesity,
                                                                                                                                                        for NAFLD  type
                                   2development.
                                      diabetes, metabolic          syndrome,       and   dyslipidemia         are  the  risk
                                                          Recently, it has been thought that the development and progression of factors   for  NAFLD         develop-
                                   ment.
                                    NAFLD    Recently,
                                                are induced it hasbybeen
                                                                       thethought       that the involving
                                                                            “multiple-hits”           development         and progression
                                                                                                                    various      factors (Figure    of NAFLD
                                                                                                                                                        3) [102,103]. are
                                   induced      by  the   “multiple-hits”      involving       various      factors   (Figure
                                    The “multiple-hits” include bioactive molecules secreted from the adipose tissue, nutri-      3) [102,103].     The    “multiple-
                                   hits”
                                    tionalinclude
                                             factors,bioactive       molecules secreted
                                                         and environmental                        from the adipose tissue, nutritional factors, and
                                                                                     factors [102].
                                   environmental factors [102].

                                    Figure 3. Multiple-hits
                                    Figure     Multiple-hitspathogenesis
                                                              pathogenesisofof
                                                                             NAFLD
                                                                               NAFLD and  NASH.
                                                                                        and     NAFLD
                                                                                            NASH.  NAFLD begins withwith
                                                                                                            begins   hepatic lipidlipid
                                                                                                                         hepatic   ac-
                                    cumulation   and insulin resistance, and progresses to NASH with the concert of various factors
                                    accumulation and insulin resistance, and progresses to NASH with the concert of various factors
                                    such as inflammation, endotoxin, organokines (adipokines and hepatokines), and oxidative stress. (:
                                    Factors related with multiple-hits).
Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC - MDPI
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW                                                                                                 6 of 30

                                    such as inflammation, endotoxin, organokines (adipokines and hepatokines), and oxidative stress.
Int. J. Mol. Sci. 2021, 22, 4495                                                                                              6 of 30
                                    (▪: Factors related with multiple-hits).

                                    2.2. Promising Therapies in NAFLD and NASH
                                    2.2. Promising Therapies in NAFLD and NASH
                                          As recently recommended pharmacotherapies, it has been reported that pioglitazone
                                          As recently recommended pharmacotherapies, it has been reported that pioglitazone and
                                    and high dosage vitamin E effectively improve the histology of patients with NASH [104–
                                    high dosage vitamin E effectively improve the histology of patients with NASH [104–106]. On
                                    106]. On the other hand, metformin does not recover liver histology of patients with
                                    the other hand, metformin does not recover liver histology of patients with NAFLD [107,108],
                                    NAFLD [107,108], and ursodeoxycholic acid (UDCA) does not improve liver histology,
                                    and ursodeoxycholic acid (UDCA) does not improve liver histology, inflammation, or fibrosis
                                    inflammation, or fibrosis of patients with NASH [109–111]. Below are some of pharmaco-
                                    of patients with NASH [109–111]. Below are some of pharmaco-therapeutic options that are
                                    therapeutic    options
                                    in clinical trials      thatbe
                                                       or could   are in clinical
                                                                    good          trialsfor
                                                                          candidates      orNASH
                                                                                             could treatment
                                                                                                    be good candidates
                                                                                                              (Figure 4). for NASH treat-
                                                                                                                          Additionally, the
                                    ment   (Figure   4). Additionally,   the metabolic    profile  and liver histology-related
                                    metabolic profile and liver histology-related efficacy of these promising drugs in humans  efficacyare
                                                                                                                                         of
                                    these  promising     drugs
                                    summarized in Table 1.      in humans    are  summarized      in Table 1.

      Figure 4. Current therapeutic targets for pharmacological treatment of NAFLD and NASH. There are no FDA-approved
      medications for
      medications  for patients
                       patients with
                                 with NAFLD/NASH
                                       NAFLD/NASH so    so far.
                                                            far. Currently,
                                                                 Currently, various
                                                                            various pharmacological
                                                                                    pharmacological therapeutic
                                                                                                    therapeutic candidates
                                                                                                                 candidates are being
                                                                                                                            are being
      applied to the clinical trials. The illustration demonstrates   the targeted  pathway and phenotype  for  treatment of patients
      applied to the clinical trials. The illustration demonstrates the targeted pathway and phenotype for treatment of patients
      with NAFLD and NASH.
      with NAFLD and NASH.

                                    2.2.1. Pioglitazone
                                           Pioglitazone
                                          Pioglitazone is one of the anti-diabetic agents of the thiazolidinediones (TZDs) class
                                    used in the management of type 2 diabetes [112]. TZDs                TZDs is is also
                                                                                                                    also known
                                                                                                                          known as “glitazones”.
                                                                                                                                        “glitazones”.
                                            are two
                                    There are    twoTZDs,
                                                       TZDs,rosiglitazone
                                                                rosiglitazoneand    and   pioglitazone,
                                                                                       pioglitazone,         which
                                                                                                          which   areare   currently
                                                                                                                       currently       approved
                                                                                                                                   approved         by
                                                                                                                                                by the
                                    the FDA
                                    FDA        as monotherapy
                                          as monotherapy              or combined
                                                                or combined              therapies
                                                                                   therapies    with with    metformin
                                                                                                       metformin             and sulfonylureas
                                                                                                                      and sulfonylureas            for
                                                                                                                                             for man-
                                    managing
                                    aging  type type    2 diabetes
                                                  2 diabetes   [113].[113].
                                                                        TZDs,  TZDs,    as insulin
                                                                                 as insulin           sensitizers,
                                                                                               sensitizers,          help regulate
                                                                                                              help regulate            glycemia
                                                                                                                                glycemia          and
                                                                                                                                            and insu-
                                    insulin
                                    lin      resistance
                                        resistance   [113].[113].  The most
                                                             The most             important
                                                                           important            advantage
                                                                                         advantage      of TZD ofisTZD
                                                                                                                     thatisitthat
                                                                                                                              doesitnot
                                                                                                                                     does   not hypo-
                                                                                                                                         cause  cause
                                    hypoglycemia
                                    glycemia           with single
                                               with single     therapy,therapy,     and there
                                                                            and there     are noare    no contraindications
                                                                                                    contraindications             for patients
                                                                                                                            for patients         with
                                                                                                                                           with renal
                                    renal disease
                                    disease  [114]. [114].
                                          TZDs regulates metabolic pathways
                                          TZDs                                pathways by binding to the nuclear transcription factor
                                    peroxisome     proliferator-activated
                                    peroxisome proliferator-activated receptor   receptor gamma
                                                                                             gamma (PPARγ)
                                                                                                        (PPARγ) and and modulating
                                                                                                                          modulating target
                                                                                                                                          target gene
                                                                                                                                                 gene
                                    expression [115]. The genes play a role in glucose metabolism, FA storage, and adipocyte
                                    expression    [115].  The  genes    play   a  role in  glucose    metabolism,      FA   storage,  and   adipocyte
                                    differentiation [116].
                                    differentiation    [116]. InIn line
                                                                    line with
                                                                            with this,
                                                                                   this, PPARγ
                                                                                          PPARγ agonists
                                                                                                     agonists increase
                                                                                                                increase glucose
                                                                                                                             glucose transporter
                                                                                                                                       transporter 44
                                    (GLUT4, also
                                    (GLUT4,    alsoknown
                                                      knownasasSLC2A4)
                                                                   SLC2A4)     expression
                                                                                  expression  andand
                                                                                                   translocation,
                                                                                                        translocation,inhibit  TNF-α,
                                                                                                                           inhibit      and enhance
                                                                                                                                    TNF-α,    and en-
                                    insulin  sensitivity   in insulin-sensitive       organs    [117,118].    On   the  other
                                    hance insulin sensitivity in insulin-sensitive organs [117,118]. On the other hand, TZD    hand,   TZD    therapy
                                    leads to leads
                                    therapy   weight  togain   as again
                                                          weight      sideas effect,
                                                                                a sidebecause
                                                                                          effect, PPARγ
                                                                                                   becausereceptors       are highlyare
                                                                                                              PPARγ receptors           expressed   in
                                                                                                                                           highly ex-
                                    adipocytes    [119].  Increases    in  fat mass   are  exclusively     limited  to  the  subcutaneous
                                    pressed in adipocytes [119]. Increases in fat mass are exclusively limited to the subcuta-                adipose
                                    depot rather
                                    neous   adiposethandepotthe rather
                                                                 visceral    spot
                                                                          than   the[117,120].
                                                                                      visceral They       can be improved
                                                                                                  spot [117,120].      They can bybe treatment
                                                                                                                                       improved  with
                                                                                                                                                    by
                                    metformin     [121,122].
                                    treatment with metformin [121,122].
                                          Recently, it was reported that the PPARγ agonist Pioglitazone has significant effects
                                    on NAFLD/NASH patients. In NASH patients, it improves liver fat accumulation and
                                    fibrosis [123,124]. In NASH patients with type 2 diabetes, it reduces hepatic steatosis, in-
Int. J. Mol. Sci. 2021, 22, 4495                                                                                          7 of 30

                                   flammation, and the serum alanine aminotransferase (ALT) and aspartate aminotransferase
                                   (AST) levels and improves the liver [125]. In rodent models, it reduces hepatic gluconeo-
                                   genesis, and improves insulin sensitivity in the liver and other peripheral tissues [126]. It
                                   also improves hepatic fibrosis [126].

                                   2.2.2. Obeticholic acid (OCA, Also Known as INT-747; FXR Agonist)
                                         OCA is a potent and selective agonist of farnesoid X receptor (FXR), a nuclear receptor
                                   that can regulate hepatic glucose and lipid metabolism, inflammation, and lipoprotein
                                   composition as well as bile acid synthesis [127,128]. In rodent models, OCA exerts the HSCs
                                   and macrophages anti-inflammatory and anti-fibrotic effects [129,130]. The transcriptional
                                   repressor small or short heterodimer partner (SHP) interacts with liver receptor homolog-1
                                   (LRH-1), a positive regulator of CYP7A1 that encodes the rate-limiting enzyme in the classic
                                   bile acid synthesis pathways, and suppresses its transcriptional activity [131]. Exposure of
                                   HSCs to FXR ligands increases the transcriptional repressor SHP expression and reduces
                                   factors associated with liver fibrosis [130]. It is thought that an FXR-SHP regulatory axis
                                   plays an important role in regulating liver fibrosis. OCA-induced FXR activity is 100-fold
                                   more potent than human natural FXR agonist, chenodeoxycholic acid (CDCA) [132]. OCA
                                   increases insulin sensitivity and reduces markers related with hepatic inflammation and
                                   fibrosis in patients with type 2 diabetes and NAFLD [133]. OCA leads to weight loss in
                                   patients with NASH, and weight loss caused by OCA is shown to exert additively beneficial
                                   effects on serum aminotransferase and liver histology [134]. Additionally, it significantly
                                   improves fibrosis in NASH patients [135]. It is one of the most promising drugs for treating
                                   NASH and is now in phase 3 clinical trials [136].

                                   2.2.3. Elafibranor (GFT505; PPARα/δ Agonist)
                                        PPARs are ligand-activated transcription factors of nuclear hormone receptor su-
                                   perfamily [137]. They are expressed in the liver, adipose tissue, skeletal muscle, heart,
                                   and kidney, and regulate metabolic pathways including β-oxidation and gluconeogen-
                                   esis [136]. There are three nuclear receptor isoforms: PPAR alpha (α), PPAR delta (δ),
                                   and PPAR gamma (γ). PPARα promotes β-oxidation, reduces TG levels, and increases
                                   high-density lipoprotein (HDL) cholesterol [138]. It also inhibits NF-kB-induced inflam-
                                   matory genes [138]. PPARα agonist, in forms such as fibric acid derivatives (fibrates), is
                                   broadly used for treatment of hypertriglycemia, whereas it has no significant effects on
                                   patients with NAFLD [139]. This is considered to be because PPARα receptors are present
                                   in other organs as well as the liver. Similar to PPARα, PPARδ increases FA oxidation and
                                   additionally reduces activation of macrophages and Kupffer cells because it is present in
                                   macrophages [140]. GW501516 is a synthetic PPARδ-specific agonist [141,142]. GW501516
                                   might be considered as a promising therapy in clinical trials because of its potent efficacy,
                                   but it has safety concerns [143].
                                        Elafibranor, also known as GFT505, is a dual PPARα/δ agonist [144]. It improves
                                   inflammation, apoptosis, and necroptosis in the NASH mouse model [145]. It reduced
                                   histopathologically hepatic steatosis and inflammation, and reduced fibrosis severity in
                                   both NAFLD/NASH and fibrosis mouse models [146,147]. It tends to reduce body weight,
                                   but not liver weight in diet-induced NAFLD/NASH rodent models [148,149]. In obese
                                   patients, it improves hepatic and peripheral insulin sensitivity [150]. Further, it inhibits
                                   proinflammatory (IL-1β, TNF-α, and F4/80) and profibrotic (transforming growth factor-β
                                   (TGF-β), tissue inhibitor of metalloproteinase 2, collagen type I, alpha 1, and collagen
                                   type I, alpha 2) markers in obese patients [151]. In addition, it decreases liver dysfunction
                                   markers such as ALT and alkaline phosphatase (ALP) [146,151]. It did not cause weight
                                   gain [144,152]. It is currently being evaluated in a pivotal phase 3 clinical trials in NASH
                                   patients [136].
Int. J. Mol. Sci. 2021, 22, 4495                                                                                             8 of 30

                                   2.2.4. Arachidyl Amido Cholanoic Acid (Aramchol)
                                         Aramchol is the liver targeted, oral stearoyl-CoA desaturase 1 (SCD1) inhibitor [153]. It
                                   is a novel fatty acid bile acid conjugate (FABACs) [153]. In rodents, aramchol affects liver fat
                                   metabolism by reducing FA synthesis and increasing β-oxidation [154,155]. Furthermore,
                                   aramchol activates cholesterol efflux by stimulating the ATP-binding cassette transporter
                                   A1 (ABCA1) [156]. In addition, it reduces inflammation and fibrosis in methionine and
                                   choline deficient (MCD) fed mice [153]. Additionally, it improves steatohepatitis and
                                   fibrosis by decreasing SCD1 levels and by regulating the transsulfuration pathway, leading
                                   to a rise in glutathione (GSH) levels and the glutathione disulfide (GSSG)/GSH redox
                                   couple to properly balance the redox environment [153]. Weight loss by aramchol treatment
                                   is known to stabilize within 1 week [153].
                                         In in a phase 2 trial of patients with NAFLD, aramchol reduces the liver fat content
                                   and improves liver histology [157]. There was no significant toxicity as determined by the
                                   circulating ALT, AST, and alkaline phosphatase levels [157]. Because it targets both the
                                   general characteristics of NASH (excessive liver fat contents, lipotoxicity, and oxidative
                                   stress) and fibrosis [153], aramchol is currently being developed for the treatment of NASH
                                   and fibrosis. It is known that there were no significant changes in the body weight of
                                   NASH patients. Phase 3 clinical trials in patients with NASH and fibrosis were initiated in
                                   2019 and are ongoing.

                                   2.2.5. Liraglutide (GLP-1R Agonist)
                                        Glucagon-like peptide-1 receptor (GLP-1R) agonists are well established as an effective
                                   medication showing promising anti-diabetic effects in both animal models and patients
                                   with type 2 diabetes [158–160]. GLP-1 is a incretin hormone that is secreted from L-cells in
                                   the distal ileum and colon [161]. It stimulates the pancreas, leading to insulin biosynthesis
                                   and insulin secretion, and reduces glucagon production [162,163]. Endogenous GLP-1
                                   is degraded within a few minutes by the dipeptidyl peptidase-4 (DPP-4) enzyme, but
                                   liraglutide works for a long time, with a half-life of 13 h [164].
                                        Exenatide, a synthetic exendin-4, was the first GLP-1R agonist approved by the FDA
                                   in 2005 for the treatment of type 2 diabetes, as monotherapy or as add-on treatment to
                                   metformin and/or sulfonylurea where control was inadequate [165].
                                        Liraglutide is the second GLP-1R agonist to be licensed the FDA in 2010 for the
                                   treatment of type 2 diabetes. It is also received FDA approval in 2020 as a treatment for
                                   obesity patients, based on its lasting weight loss benefits [166,167]. It has cardiovascular
                                   safety in treatment for weight management [168]. Liraglutide-induced anorexia is also
                                   related with glutamatergic POMC neuron, leading to weight loss [169]. In patients with
                                   NAFLD and NASH, it decreases liver fat contents and improves histological resolution
                                   and serum liver enzyme levels without worsening fibrosis [170,171]. It is thought that
                                   the effect of liraglutide on weight loss and reduced cardiovascular risk is critical for
                                   treatment of NAFLD because the development of NAFLD is based on lipotoxicity and
                                   insulin resistance [171,172]. As studies associated with NAFLD and NASH in rodents
                                   showed, liraglutide protects pancreatic β-cells from apoptosis through AKT-mediated
                                   survival signaling [173]. It improves insulin sensitivity by activating AMP-activated protein
                                   kinase (AMPK) and reduces liver steatosis by modulating lipid transport, β-oxidation,
                                   DNL, and autophagy [174–176].

                                   2.2.6. Selonsertib (ASK1 Inhibitor)
                                        Ballooned hepatocytes, implicating activation of the apoptotic pathway, are a hall
                                   marker of NASH and fibrosis progression [177,178]. Selonsertib is a first-in-class inhibitor
                                   of the apoptosis signal regulating kinase 1(ASK1) [179]. It inhibits the phosphorylation
                                   and activation of ASK1 by binding to the catalytic kinase domain of ASK1. Recently, it
                                   has proposed as therapeutic potential for fibrotic diseases. In mouse models, ASK1, a
                                   serine/threonine signaling kinase, causes phosphorylation of p38 mitogen-activated kinase
                                   and c-Jun N-terminal kinase (JNK), leading to activation of stress response pathways that
Int. J. Mol. Sci. 2021, 22, 4495                                                                                                                     9 of 30

                                          aggravate hepatic inflammation, apoptosis, and fibrosis [180–182]. In mouse models of
                                          NASH, it significantly improves not only liver steatosis and fibrosis associated with NASH
                                          but also cholesterol, bile acid, and lipid metabolism [180]. In phase 2 clinical trials of
                                          patients with NASH and stage 2–3 fibrosis, it has been shown to prevent inflammation,
                                          fibrosis, excessive apoptosis, and progression to cirrhosis [183]. On the other hand, phase 3
                                          clinical trials of patients with NASH and advanced fibrosis were found to improve liver
                                          histology, but did not affect fibrosis regression [184,185]

                                          2.2.7. Simtuzumab (SIM, G6624)
                                                SIM is a monoclonal antibody targeting the lysyl oxidase-like 2 (LOXL2) enzyme that
                                          catalyzes the crosslinkage of collagen and elastin, leading to remodeling of the extracel-
                                          lular matrix [186,187]. SIM binds to LOXL2 and inhibits its enzymatic activity [188]. As
                                          a result, it inhibits synthesis of growth factors including connective tissue growth fac-
                                          tor (CTGF/CCN2) and TGFβ1, and reduces liver fibrosis [189]. In a mouse model with
                                          advanced fibrosis, SIM has an additive effect in combination with ASK1 inhibitor [183].
                                          However, in phase 2b clinical trials of subjects with advanced fibrosis induced by NASH,
                                          it was no effect on improving fibrosis and cirrhosis confirmed by hepatic collagen con-
                                          tent [190].

                                          2.2.8. Cenicriviroc (CVC; Dual CCR2/CCR5 Antagonist)
                                                Liver inflammation is closely associated with chemokines that regulate activities
                                          and migration of hepatocytes and immune cells [191]. The C-C chemokine receptors
                                          CCR2 and CCR5 with their respective ligands (CCL2 and CLL3-5) are associated with
                                          the pathogenesis of liver inflammation and fibrosis for the development of NAFLD and
                                          NASH [191–193]. CCR2 and its ligand CCL2 enhances hepatic steatosis, macrophage
                                          accumulation, inflammation, and fibrosis [191]. Activated HSCs, a contributor for fibrosis,
                                          secretes CCL5. CCL5 exerts profibrotic activity in hepatocytes via its receptors CCR5 and
                                          induces lipid accumulation and pro-inflammatory factors [192].
                                                CVC is a novel and potent antagonist of CCR2 and CCR5 that is currently in clinical
                                          development for treatment of liver fibrosis in patients with NASH [194,195]. CVC reduces
                                          levels of inflammation markers including IL-1β and IL-6 and exerts anti-fibrotic activi-
                                          ties [194,195]. It received Fast Track designation by the FDA in 2015 as a promising therapy
                                          for NASH and liver fibrosis. In the phase 2b study of subjects with NASH and stage 2–3
                                          fibrosis, CVC has shown improvement in liver fibrosis without worsening NASH [196].
                                          Currently, phase 3 clinical trials are ongoing to evaluate and confirm the efficacy and safety
                                          of CVC for the treatment of liver fibrosis in patients with NASH [197].

                                          2.3. Diagnostic and Therapeutic Targets in NAFLD and NASH: Adipokines
                                               Recently, it has been believed that NAFLD and NASH are caused by the multiple
                                          factors [102,103]. Among them, we will focus on adipokines secreted from adipose tissues
                                          that provide FA as the major source for NAFLD development [48]. Several adipokines
                                          are involved in the pathogenesis and progression of NAFLD [198]. Leptin, resistin, and
                                          visfatin play a role in NAFLD development and progression to NASH [199–204]. On
                                          the other hand, adiponectin, irisin, and ghrelin exert beneficial effects on NAFLD and
                                          NASH [205–211]. Pharmacological agents that affect liver histology and pathophysiology
                                          could be influential in theses adipokine levels. It suggests that adipokines can be attractive
                                          targets for treatment and can be biomarkers for prediction of NAFLD severity (Figure 5).
                                          Adipokines can also play an active role in the development of HCC.

                                             Table 1. Summary of promising drugs for NAFLD/NASH.

                                                                                                            Clinical Stage
      Name of Drug            Mechanism of Action       Metabolic Profile        Liver Histology                                           Ref.
                                                                                                            (Title of Trial)
                                                        Insulin sensitivity(↑)           Steatosis(↓)
                                                              Hepatic TG(↓)            Ballooning(↓)
       Pioglitazone                PPAR γ agonist                     ALT(↓)        Inflammation(↓)     Phase 4 trial, 2008–2014   [104,105,125,212,213]
                                                                      AST(↓)              Fibrosis(↓)
                                                                       BW(↑)                 NAS(↓)
                                                        Insulin sensitivity(↑)
                                                                      ALT(↓)             Steatosis(↓)
                                                                                                        Phase 3 trial, ongoing
                                                                      AST(↓)           Ballooning(↓)
                                                                                                             since 2017
  Obeticholic acid (OCA)            FXR agonist                       ALP(↑)        Inflammation(↓)                                      [214,215]
                                                                                                          (REGENERATE,
                                                                   HDL-C(↓)               Fibrosis(↓)
                                                                                                             REVERSE)
                                                                   LDL-C(↑)                  NAS(↓)
                                                                       BW(↓)
Int. J. Mol. Sci. 2021, 22, 4495                                                                                                                       10 of 30

                                                                        Table 1. Cont.

                                                                                                                  Clinical Stage
      Name of Drug            Mechanism of Action        Metabolic Profile            Liver Histology                                        Ref.
                                                                                                                  (Title of Trial)
                                                          Insulin sensitivity(↑)
                                                                 Plasma TG(↓)
                                                                                               Steatosis(↓)   Phase 3 trial, ongoing
                                                                        ALT(↓)
        Elafibranor           Dual PPARα/δ agonist                                        Inflammation(↓)          since 2016            [144,150,151]
                                                                         AST(-)
                                                                                                Fibrosis(↓)      (RESOLVE-IT)
                                                                        ALP(↓)
                                                                         BW(-)
                                                          Insulin sensitivity(↑)
                                                                Hepatic TG(↓)
                                                                                               Steatosis(↓)   Phase 3 trial, ongoing
        Aramchol                    SCD1 inhibitor                       ALT(-)                                                              [157]
                                                                                                Fibrosis(↓)   since 2019 (ARMOR)
                                                                         AST(-)
                                                                         BW(-)
                                                          Insulin sensitivity(↑)
                                                                Hepatic TG(↓)                                 Phase 3 trial, ongoing
                                                                                              Steatosis(↓)
        Liraglutide                 GLP-1R agonist                      ALT(↓)                                     since 2014            [164,216,217]
                                                                                            Ballooning(↓)
                                                                         AST(-)                                 (CGH-LiNASH)
                                                                         BW(↓)
                                                                                               Steatosis(↓)
                                                                        ALT(↓)               Ballooning(↓)     Phase 3 trial, ongoing
        Selonsertib                 ASK1 inhibitor                      AST(↓)            Inflammation(↓)     since 2019 (STELLAR3,          [93]
                                                                         BW(-)                  Fibrosis(↓)         STELLAR4)
                                                                                                   NAS(↓)
                                                                        ALT(-)
                                   LOXL2 monoclonal
    Simtuzumab (SIM)                                                    AST(-)                  Fibrosis(-)   Phase 2 trial, 2012–2017     [218,219]
                                      antibody
                                                                        BW(-)
                                                                        ALT(-)
                                Dual CCR2/CCR5                                            Inflammation(↓)     Phase 3 trial, ongoing
    Cenicriviroc (CVC)Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW     AST(-)                                                               [196]                11
                                     antagonist                                                 Fibrosis(↓)   since 2017 (AURORA)
                                                                        BW(-)

                                                      ((↑): increase, (↓): decrease, (-): no significant).

                             Figure 5. Adipokines
                                       Figure 5. as   diagnostic as
                                                   Adipokines     markers   and therapeutic
                                                                     diagnostic  markers andtargets in NAFLD
                                                                                                 therapeutic     and NASH.
                                                                                                               targets      Adipokines
                                                                                                                       in NAFLD         that are secreted
                                                                                                                                  and NASH.
                             from adipose tissues are
                                       Adipokines     classified
                                                    that         into anti-inflammatory
                                                         are secreted                    adipokines
                                                                       from adipose tissues           and pro-inflammatory
                                                                                             are classified                  adipokines.
                                                                                                            into anti-inflammatory       Anti-inflamma
                                                                                                                                   adipokines
                             tory adipokines including adiponectin, irisin, and ghrelin inhibit the development and progression of NAFLD and NASH
                                       and pro-inflammatory adipokines. Anti-inflammatory adipokines including adiponectin, irisin, and
                             whereas pro-inflammatory adipokines including leptin, resistin, and visfatin promote the development and progression
                                       ghrelin inhibit the development and progression of NAFLD and NASH, whereas pro-inflammatory
                             of NAFLD and NASH.
                                       adipokines including leptin, resistin, and visfatin promote the development and progression of
                                       NAFLD and NASH.   2.3.1. Adiponectin
                                           2.3.1. Adiponectin Adiponectin is an important adipokine that can inhibit NAFLD development. C
                                                          lating
                                                 Adiponectin is anadiponectin levels werethat
                                                                   important adipokine     decreased  in patients
                                                                                              can inhibit NAFLDwith    NAFLD and
                                                                                                                   development.      NASH [220–
                                                                                                                                   Circu-
                                           lating adiponectin levels were decreased in patients with NAFLD and NASH [220–222].inflamma
                                                          These   are inversely correlated with the  severity of  hepatic steatosis and
                                                          Pioglitazone, an anti-diabetic drug of thiazolidinedione-type, improves liver histo
                                                          and increases adiponectin levels in patients with NASH [104,125]. However, metfo
                                                          the most commonly used anti-diabetic medication, has no significant effects on the
                                                          histology of patients with NAFLD and NASH, and reduces adiponectin l
                                                          [107,108,223]. Vitamin E is a potent antioxidant that protects our cells against oxid
Int. J. Mol. Sci. 2021, 22, 4495                                                                                            11 of 30

                                   These are inversely correlated with the severity of hepatic steatosis and inflammation.
                                   Pioglitazone, an anti-diabetic drug of thiazolidinedione-type, improves liver histology
                                   and increases adiponectin levels in patients with NASH [104,125]. However, metformin,
                                   the most commonly used anti-diabetic medication, has no significant effects on the liver
                                   histology of patients with NAFLD and NASH, and reduces adiponectin levels [107,108,223].
                                   Vitamin E is a potent antioxidant that protects our cells against oxidative stress [224]. It is
                                   an alternative medicine that is recommended in NAFLD and NASH. Vitamin E improves
                                   liver histology and shows some beneficial effects in non-diabetic patient with NASH,
                                   and also seems to increase adiponectin levels [225,226]. However, it has been found to
                                   be ineffective alone in NASH patients with type 2 diabetes [226,227]. In mouse models,
                                   adiponectin suppresses hepatic lipid accumulation by enhancing FA oxidation and re-
                                   ducing DNL [206,228,229]. It exerts anti-inflammation, anti-fibrotic, and anti-apoptotic
                                   effects [229]. Administration of adiponectin improves hepatic steatosis and inflamma-
                                   tion [228,229]. Additionally, adiponectin expression is inversely correlated with tumor size
                                   and local recurrence [230,231].

                                   2.3.2. Leptin
                                        Leptin is an appetite-suppressing hormone secreted by fat cells. It regulates food in-
                                   take, body fat, and insulin sensitivity [232]. In animal models, it is thought that it improves
                                   lipid metabolism in non-adipose tissues [233]. In the liver, however, it exacerbates hepatic
                                   insulin resistance, which results in liver steatosis. It also enhances liver fibrosis [199,233].
                                   Leptin administration can enhance pro-inflammatory and fibrogenic responses in the liver
                                   via procollagen I and TGFβ1 [234]. In humans, however, its effects are unclear. Circulating
                                   leptin levels are increased in patients with NASH [235,236]. Leptin levels are positively
                                   correlated with steatosis severity, whereas it is unclear between leptin levels and the pro-
                                   gression of inflammation and fibrosis [235–238]. Leptin expression is positively correlated
                                   with cell proliferation in HCC, as confirmed by proliferation marker protein Ki67 [231].

                                   2.3.3. Resistin
                                        Resistin is a proinflammatory adipocyte-derived mediator of hepatic insulin resis-
                                   tance [239,240]. It is also expressed in liver cells. Resistin is associated with hepatic
                                   lipogenesis and liver fibrosis [241]. Circulating resistin levels are increased in patients with
                                   NAFLD and NASH, and circulating resistin levels in patients with NAFLD are related to the
                                   severity of steatosis, inflammation, and fibrosis [202,242,243]. Increased resistin levels are
                                   thought to be associated with insulin resistance. In individuals with NAFLD, pioglitazone
                                   treatment improves insulin sensitivity, and decreases plasma resistin levels [244].

                                   2.3.4. Ghrelin
                                        Ghrelin is an anti-inflammatory adipokine. It is an endogenous ligand for growth
                                   hormone secretagog receptor with a peptide structure that contains 28 amino acids [245]. In
                                   patients with NAFLD, lower ghrelin levels are associated with insulin resistance [246,247].
                                   Plasma ghrelin levels are significantly correlated with liver function. However, ghrelin
                                   is not affected by pioglitazone as one of insulin sentizers [45]. During and after NAFLD
                                   development, ghrelin administration improves hepatic lipid metabolism, inflammation,
                                   oxidative stress, and apoptosis [210]. In mouse models, ghrelin reduces the TG content and
                                   the cytokins TNF-α and IL-6, and attenuates lipotoxicity through autophagy sitimulation
                                   and NF-kB inhibition [248]. Collectively, ghrelin could be a biomarker for diagnosis and a
                                   therapeutic target for treatment of NALFD.

                                   2.3.5. Irisin
                                         Irisin is a myokine secreted from skeletal muscle upon shivering and exercise stimula-
                                   tion [249]. Fibronectin type III domain containing 5 precursors (FNDC5) is the precursor
                                   of irisin. FNDC5/irisin promotes the thermogenic program in adipose tisseus through
                                   ERK and p38 pathways [250]. It improves glucose homeostasis and insulin resistance, and
Int. J. Mol. Sci. 2021, 22, 4495                                                                                            12 of 30

                                   induces weight loss [251]. Recently, FNDC5/irisin was induced during adipocyte differen-
                                   tiation, and can be over-secreted from human obese visceral (VAT) and subcutaneous (SAT)
                                   adipose tissues [252]. It is thought of as a compensatory effect. In line with this, circulating
                                   irisin levels are increased in patients with NAFLD, and are positively related with portal
                                   inflammation [218]. They are also believed to act as a compensatory effect.

                                   2.3.6. Visfatin
                                        Visfatin is one of the proinflammatory adipokines. Serum visfatin levels are raised in
                                   type 2 diabetes and insulin resistant conditions [253,254]. Circulating visfatin levels are also
                                   increased in patients with NAFLD, and are associated with the severity of hepatic steatosis
                                   and fibrosis [204,255]. However, they are not affected by insulin sensitizers including
                                   pioglitazone, rosiglitazone, and metformin [256,257].

                                   3. NAFLD/NASH-Derived HCC
                                   3.1. The Pathogenesis of NAFLD/NASH-Derived HCC
                                         HCC is the third most common cause of cancer-related mortality [258]. NAFLD and
                                   NASH-related HCC is the fastest growing indication for liver transplantation [259,260].
                                   Cirrhosis is only present in approximately 60% of patients with NAFLD and NASH-
                                   associated HCC [259]. This suggests that HCC can be induced from NAFLD/NASH
                                   without cirrhosis. Therefore, it is thought that “inflammatory factors” will also play a
                                   critical role in NAFLD/NASH-derived HCC.

                                   3.1.1. Gut-Derived Endotoxin
                                        As mentioned above, gut-derived endotoxins as alternative inflammatory factors play
                                   an important role in the development of NAFLD and NASH. The levels of LPS, known as
                                   endotoxins, are also increased in portal and peripheral veins of patients with HCC [261].
                                   They significantly promote the invasive potential and induce the epithelial-mesenchymal
                                   transition (EMT), although they also inhibit tumor growth [262]. LPS activates JNK and
                                   MAPK via TLR4 in HCC cells, whereas inhibition of JNK/MAPK significantly reduces
                                   EMT occurrence [262]. Therefor, the LPS-TLR4 signaling could be one of the promising
                                   pathways regulating the progression from NAFLD to NASH to HCC [263].

                                   3.1.2. Adipokines
                                         Adipokines are inflammatory factors related with HCC development. Adiponectin
                                   expression in human HCC is inversely correlated with tumor size [230]. It enhances
                                   phosphorylation of c-Jun N-terminal kinase (JNK) and activates caspase-3, leading to
                                   apoptosis in HCC [230]. Inhibition of JNK phosphorylation prevents anti-apoptotic effects
                                   of adiponectin [230]. Adiponectin exerts chmoprotective and hepatoprotective effects
                                   via sulfatase2 (SULF2) in HCC [264]. Loss of adiponectin promotes fibrosis and HCC
                                   progression in a cholin-deficient NASH mouse model [265]. On the other hand, high levels
                                   of circulating adiponectin make it possible to predict the consecutive development of HCC
                                   and poor HCC survival [266,267]. Further, adiponectin inhibits the oncogenic effects of
                                   leptin on cell proliferation, migration, and invasion in HCC [231].
                                         Leptin expression is increased in both hepatoma tissues and cell lines [268]. Regulatory
                                   T-cells (Tregs), effector CD4(+), and CD8(+) T-cells stimulate expression of the leptin
                                   receptor (LEPR) in the liver after HCC induction [268]. Macrophage and dendritic cells
                                   upregulate LEPR expression on the T-cell. Leptin inhibits Treg activation and function [268].
                                   Increased leptin expression in HCC is associated with the expression of human telomerase
                                   reverse transcriptase (hTERT) [269]. Leptin might play a critical role in obesity-related
                                   tumorigenesis. Adipokines including adiponectin and leptin represent key players in
                                   obesity-related disorders and might be involved in the pathogenesis of NAFLD and HCC.
Int. J. Mol. Sci. 2021, 22, 4495                                                                                                      13 of 30

                                   3.2. Diagnostic and Therapeutic Targets in NAFLD/NASH-Derived HCC: Hepatokines
                                             The liver is a secretory organ that releases specific cytokines, termed hepatokines [43].
                                       Adipose tissues in NAFLD, characterized by hepatic TG accumulation, play a critical
                                       role in promoting FFA uptake into the liver through lipolysis [48]. Therefore, the role of
                                       adipokines from adipose tissues, which provide the major energy source for the devel-
                                       opment of NAFLD, will be very important in the liver. On the other hand, lipid droplet
                                       accumulation itself does not affect inflammation and is considered as simple steatosis.
                                       The progression from NAFLD to NASH to HCC needs additional factors such as oxida-
                                       tive stress, mitochondrial dysfunction, and ER-stress [75,270,271]. Another important
                                       factor driving NASH in simple steatosis is free non-esterified cholesterol and its oxidized
                                       derivatives [272–274]. They are cytotoxic and exert synergistic effects with TNF, which is
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
                                       markedly increased in patients with NASH [274]. Therefore, hepatokines secreted from   14 ofthe
                                                                                                                                    30
                                       liver might exert a more potent ability in the progression of NAFLD and NASH to HCC
                                       (Figure 6).

      Figure 6. Hepatokines that
                              that are
                                   are secreted
                                        secretedfrom
                                                 fromthe
                                                       theliver
                                                            liverare
                                                                  areclosely
                                                                       closelyassociated
                                                                               associatedwith
                                                                                           withthe
                                                                                                 theprogression
                                                                                                     progressionfrom NAFLD
                                                                                                                  from NAFLDto to
                                                                                                                               NASH
                                                                                                                                  NASHto
      to HCC.
      HCC.     Hepatokines
            Hepatokines     including
                          including      Fetuin-A,
                                       Fetuin-A,    Fetuin-B,RBP4,
                                                  Fetuin-B,     RBP4,and
                                                                       andFGF19
                                                                            FGF19play
                                                                                    playan
                                                                                         animportant
                                                                                            importantrolerolein
                                                                                                              inNAFLD
                                                                                                                NAFLD and
                                                                                                                       and NASH. They
      are associated with
      are associated with hepatic
                           hepatic lipid
                                    lipid accumulation,
                                           accumulation, insulin
                                                           insulin resistance,
                                                                     resistance, and
                                                                                 and inflammatory
                                                                                      inflammatory signaling
                                                                                                       signaling pathways.
                                                                                                                 pathways. Additionally,
                                                                                                                           Additionally,
      ANGPTL4    and 8 tend  to function   in opposite ways    in HCC    tumorigenesis.
      ANGPTL4 and 8 tend to function in opposite ways in HCC tumorigenesis.

                                                                  (Fetuin-A and Fetuin-B)
                                          α2-HS-Glycoprotein (Fetuin-A
                                   3.2.1. α2-HS-Glycoprotein
                                         Fetuin-A, one
                                        Fetuin-A,   one of
                                                         of the liver secreted glycoproteins, is known as the first hepatokine
                                   shown
                                   shown    to associated with metabolic diseases [275,276]. Fetuin-A is positively associated
                                   with hepatic steatosis and insulin resistance [277–279]. Its levels are increased in patients
                                   with NAFLD,
                                   with   NAFLD, NASH,
                                                   NASH, and and type
                                                                   type 22 diabetes
                                                                           diabetes [280,281].   As an
                                                                                     [280,281]. As   an important   source of
                                                                                                        important source      of NAFLD
                                                                                                                                 NAFLD
                                   development, FFA
                                   development,     FFAenhances
                                                         enhancespro-inflammatory
                                                                      pro-inflammatoryFetuin-A
                                                                                            Fetuin-A   expression
                                                                                                     expression  [7].[7].  FFA-induced
                                                                                                                      FFA-induced    Fe-
                                   Fetuin-A
                                   tuin-A     functions
                                           functions  asas
                                                         anan  endogenousligand
                                                             endogenous       ligandofofToll-like
                                                                                         Toll-likereceptor
                                                                                                   receptor44(TLR4),
                                                                                                              (TLR4), and
                                                                                                                        and exacerbates
                                                                                                                             exacerbates
                                   lipid-mediated insulin
                                   lipid-mediated    insulin resistance
                                                              resistance [282,283].
                                                                           [282,283]. FFA
                                                                                      FFA can
                                                                                           can also
                                                                                                also enhances   NF-kB recruitment
                                                                                                     enhances NF-kB       recruitment to
                                                                                                                                      to
                                   the Fetuin-A promoter and increases synthesis and the secretion of Fetuin-A in primary
                                   hepatocytes [284]. Pioglitazone significantlly suppresses serum Fetuin-A levels in patients
                                   with type 2 diabetes [285]. Pioglitazone inhibits mRNA and protein levels of hepatic Fe-
                                   tuin-A, and oral administration of pioglitazone in mice partially ameliorates insulin re-
                                   sistance with decreases on hepatic fetuin-A expression [286]. These data suggest that Fe-
You can also read